Open Access

Downregulation of miR‑193a‑3p via targeting cyclin D1 in thyroid cancer

  • Authors:
    • Xiao‑Jiao Li
    • Rong Wen
    • Dong‑Yue Wen
    • Peng Lin
    • Deng‑Hua Pan
    • Li‑Jie Zhang
    • Yu He
    • Lin Shi
    • Yong‑Ying Qin
    • Yun‑Hui Lai
    • Jing‑Ni Lai
    • Jun‑Lin Yang
    • Qin‑Qiao Lai
    • Jun Wang
    • Jun Ma
    • Hong Yang
    • Yu‑Yan Pang
  • View Affiliations

  • Published online on: July 8, 2020     https://doi.org/10.3892/mmr.2020.11310
  • Pages: 2199-2218
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Thyroid cancer (TC) is a frequently occurring malignant tumor with a rising steadily incidence. microRNA (miRNA/miR)‑193a‑3p is an miRNA that is associated with tumors, playing a crucial role in the genesis and progression of various cancers. However, the expression levels of miR‑193a‑3p and its molecular mechanisms in TC remain to be elucidated. The present study aimed to probe the expression of miR‑193a‑3p and its clinical significance in TC, including its underlying molecular mechanisms. Microarray and RNA sequencing data gathered from three major databases, specifically Gene Expression Omnibus (GEO), ArrayExpress and The Cancer Genome Atlas (TCGA) databases, and the relevant data from the literature were used to examine miR‑193a‑3p expression. Meta‑analysis was also conducted to evaluate the association between clinicopathological parameters and miR‑193a‑3p in 510 TC and 59 normal samples from the TCGA database. miRWalk 3.0, and the TCGA and GEO databases were used to predict the candidate target genes of miR‑193a‑3p. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes and protein‑protein interaction network enrichment analyses were conducted by using the predicted candidate target genes to investigate the underlying carcinogenic mechanisms. A dual luciferase assay was performed to validate the targeting regulatory association between the most important hub gene cyclin D1 (CCND1) and miR‑193a‑3p. miR‑193a‑3p expression was considerably downregulated in TC compared with in the non‑cancer controls (P<0.001). The area under the curve of the summary receiver operating characteristic was 0.80. Downregulation of miR‑193a‑3p was also significantly associated with age, sex and metastasis (P=0.020, 0.044 and 0.048, respectively). Bioinformatics analysis indicated that a low miR‑193a‑3p expression may augment CCND1 expression to affect the biological processes of TC. In addition, CCND1, as a straightforward target, was validated through a dual luciferase assay. miR‑193a‑3p and CCND1 may serve as prognostic biomarkers of TC. Finally, miR‑193a‑3p may possess a crucial role in the genesis and progression of TC by altering the CCND1 expression.

Introduction

Thyroid cancer (TC) is a frequently occurring malignant tumor with steadily increasing incidence (15). In 2018, the American Cancer Society predicted that there would be 53,990 newly diagnosed patients with TC that year (6). Based on histopathological examination, TC is divided into 4 subtypes: Anaplastic thyroid carcinoma, papillary thyroid carcinoma, medullary thyroid cancer and follicular thyroid cancer (79). Different hypotypes of TC lead to various clinical outcomes. A number of studies have reported that intricate biological processes participating in the interactions between polygenes may contribute to the occurrence and progression of TC (1012). Previous studies have suggested that hsa-miR-200a-5p and hsa-miR-181a-2-3p could be applied for latent diagnostic and prognostic biomarkers of TC (13,14). Nevertheless, the clinical characteristics of molecular profiles in TC have yet to be elucidated. Therefore, to further understand the molecular mechanisms of TC, an multi-method study may offer novel insight for TC prevention and treatment (15).

MicroRNAs (miRNAs/miRs) contain ~19–25 nucleotides and are endogenous non-coding RNAs with gene-regulating functions (1619). miRNAs are critical in the biological processes of diversified human carcinomas. For instance, they participate in extensive biological actions, including cell proliferation, invasion and apoptosis (18,2023). Previous studies have demonstrated that miR-193a-3p is a neoplasm suppressor in various carcinomas, including hepatocellular cancer (24), gastric cancer (25,26), lung carcinoma (27), breast carcinoma (28) and colorectal carcinoma (29,30). To the best of the authors' knowledge, thus far only one study has investigated the association between miR-193a-3p and medullary thyroid carcinoma (31); however, a limitation of this study was the small sample size, which prevented a comprehensive description and analysis of TC. To further comprehend the potential molecular mechanisms, it is necessary to examine miR-193a-3p expression in TC using a larger volume of data.

Cyclin D1 (CCND1), a member of the highly conserved cyclin family, can function as a modulator of cyclin-dependent kinase (CDK)4 or CDK6, and has an important role in adjusting the progression of the cell cycle (32,33). Previous studies revealed that CCND1 has a crucial role in promoting proliferation, migration and tumor metastasis (34,35) in cancers such as lung cancer (33), gastric cancer (36) and renal cell cancer (37). CCND1 has been reported to exhibit a high expression in TC tissues and to be associated with invasive behavior (38).

The present study examined TC specimen information from The Cancer Genome Atlas (TCGA) to assess the expression levels of miR-193a-3p and CCND1, and their association with clinical parameters. The data of miR-193a-3p and CCND1 were extracted and validated via ArrayExpress databases and the Gene Expression Omnibus (GEO). Then, the candidate target genes of miR-193a-3p were identified using miRWalk 3.0 and overlapped with the upregulated genes in TC identified using TCGA database. The miR-193a-3p candidate target genes were further investigated using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and protein-protein interaction (PPI) network enrichment analyses. According to the bioinformatics analysis of the present study, a hub gene was found in TC, and the targeting regulatory relations between the hub gene and miR-193a-3p were confirmed by a dual luciferase assay. By performing a meta-analysis and bioinformatics analysis, the present study revealed the latent molecular mechanisms of miR-193a-3p, as well as its associations with the identified hub gene, which may lead to TC.

Materials and methods

miR-193a-3p expression in TC from public databases and the literature

TCGA is a pool of molecular datasets for at least 30 types of cancers, including TC (39). The present study explored TCGA to determine miR-193a-3p expression in TC tissues. miRNA expression data of 510 TC and 59 normal samples were acquired, along with the accompanying clinical information, from TCGA, up to December 1, 2018 (40). Chip datasets (GSE40807, GSE62054 and GSE73182) from GEO were also searched to examine the profiling of miR-193a-3p expression in TC (4143). The following keywords were used: (papillary OR medullary OR follicular OR anaplastic OR thyroid) AND (carcinoma OR cancer OR adenocarcinoma OR tumor OR neoplas* OR malignan*) AND (microRNA OR miR OR miRNA), where ‘*’ refers to a wildcard search. The public database of microarray gene expression, ArrayExpress (E-MTAB-736) (44) was also used. In addition, literature that included miR-193a-3p expression in TC was searched for in 13 online databases: Wiley Online Library (https://onlinelibrary.wiley.com), Ovid (https://www.ovid.com), LILACS (https://lilacs.bvsalud.org), Web of Science (http://www.isiknowledge.com), PubMed (https://www.ncbi.nlm.nih.gov/pubmed), Science Direct (https://www.sciencedirect.com), Cochrane Central Register of Controlled Trials (https://www.cochranelibrary.com), EMBASE (https://www.embase.com), Google Scholar (https://scholar.google.com), Chong Qing VIP (http://lib.cqvip.com), Wan Fang (http://www.wanfangdata.com), Chinese CNKI (https://www.cnki.net) and the China Biology Medicine Disc (http://www.sinomed.ac.cn). The studies needed to meet the following requirements for inclusion: i) The miR-193a-3p expression data in TC could be tested in humans; and ii) the relevant information of miR-193a-3p could be abstracted from the literature. All expression data and clinical information connected to miR-193a-3p were extracted.

Comprehensive statistical analysis of TCGA, GEO, ArrayExpress and the literature

The expression levels of both TC and non-cancerous samples were visualized using scatter diagrams and receiver operating characteristic (ROC) curves created using GraphPad Prism 6.0 (GraphPad Software, Inc.). Student's t-test was performed to compare the expression of TC and non-cancer samples, as well as the association between clinicopathological parameters and miR-193a-3p from TCGA database. One-way analysis of variance was used for three or more groups. All miR-193a-3p expression data abstracted from various databases were normalized to log2, and P<0.05 was considered to indicate a statistically significant difference. An all-sided analysis was performed by employing STATA 12.0 software (StataCorp LLC). The pooled standard mean difference (SMD) was applied to evaluate miR-193a-3p expression. χ2 and I2 tests were computed to assess the heterogeneity within the meta-analysis. The Mantel-Haenszel fixed-effects model was selected if there was no prominent heterogeneity (I2<50%). Otherwise, a random-effects model was applied (I2>50%) (45); meanwhile, heterogeneity analysis was performed to discover the sources of heterogeneity. Subsequently, a summary (s)ROC was created to indicate the ability of miR-193a-3p to identify TC compared with a normal sample (4648).

Prediction of miR-193a-3p candidate target genes and their molecular functions

To further search for the latent molecular functions of miR-193a-3p in TC, bioinformatics analysis was performed. First, the target genes of miR-193a-3p were predicted using 12 platforms [miRWalk 2.0 (version 2; http://zmf.umm.uni-heidelberg.de/apps/zmf/mirwalk2), PITA (version 6; http://genie.weizmann.ac.il/pubs/mir07/mir07_exe.html), PicTar2 (https://pictar.mdc-berlin.de), RNAhybrid 2.1 (https://bibiserv.cebitec.uni-bielefeld.de/rnahybrid), MicroT4 v4.0 (version 5; http://diana.imis.athena-innovation.gr), miRDB 4.0 (http://www.mirdb.org), miRanda-rel2010 (http://www.microrna.org/microrna/getDownloads.do), TargetScan 7.2 (version 7.2; http://www.targetscan.org), RNA22 v2 (https://cm. jefferson.edu/rna22-full-sets-of-predictions), miRBridge (http://www.ncbi.nlm.nih.gov/pubmed/?term=20385095), miRNAMap (ftp://mirnamap.mbc.nctu.edu.tw/miRNAMap2) and miRMap (https://mirmap.ezlab.org)]. The predicted genes exhibited in the 12 platforms were selected: 3,984 candidate target genes. Log2 conversion and normalization was performed for the gene chips and RNA sequencing from the GEO and TCGA databases. Differentially expressed genes in microarrays and RNA sequencing data were also screened out by the limma package (version 3.42.2) of R language (R version 3.6.2; http://www.r-project.org) (49). Integration of differentially expressed genes identified from gene chips and RNA sequencing was conducted via RobustRankAggreg (version 1.1; https://cran.r-project.org/web/packages/RobustRankAggreg). The overexpressed genes in TC from the TCGA and GEO databases were calculated by selecting the following criteria: P-adj<0.05 and any upregulated genes with a log2-fold change (FC)>1. The candidate target genes obtained with the online prediction tools in TCGA and GEO were then overlapped and presented as Venn diagrams (50). Then, the overlapping target genes were used to study molecular mechanisms via GO (51), KEGG pathway (52), Disease Ontology (DO) (53) and PPI network analyses. Bioinformatics analyses were implemented using clusterprofiler package (version 3.10) of R language (54). The ggplot2 package (version 3.3.0) from R language was utilized to visualize the results of the functional analysis (55). STRING 11.0 (http://string.embl.de) was also used to generate a PPI network, which revealed the connection among the overlapping related genes (56).

Expression of CCND1 in TC samples from the TCGA and GEO databases

The clinical value of CCND1, whose expression data was extracted from GEO and TCGA, was further explored. CCND1 expression data and its clinical information were acquired from TCGA. The GEO database was also mined to obtain chip datasets from TC samples using the following keywords: (papillary OR medullary OR follicular OR anaplastic OR thyroid) AND (carcinoma OR tumor OR cancer OR adenocarcinoma OR neoplas* OR malignan*) AND (Cyclin D1 OR G1/S-Specific Cyclin-D1 OR PRAD1 Protein OR B-Cell CLL/Lymphoma 1 OR U21B31 OR BCL-1 Oncogene OR BCL1 OR PRAD1 Oncogene OR Parathyroid Adenomatosis 1 OR D11S287E OR B-Cell Lymphoma 1 Protein). Filtering condition: Series [Entry type], Homo sapiens [Organism]. A total of 13 datasets were obtained: GSE3678, GSE6004, GSE6339, GSE9115, GSE27155, GSE29265, GSE33630, GSE35570, GSE50901, GSE53072, GSE53157, GSE58689 and GSE65144 (5767). Statistical analysis of CCND1 was also performed. The methods used were the same as those used above for evaluating the expression of miR-193a-3p.

Cell culture and transfection

The 293 cell line was obtained from Huzhou Hippo Biotechnology Co., Ltd. and cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin/streptomycin. Cells were cultured in an incubator at 37°C with 5% CO2. miR-193a-3p mimic (5′-AACUGGCCUACAAAGUCCCAGU-3′), miR-146b-5p mimic (5′-UGAGAACUGAAUUCCAUAGGCUG-3′) and mimic control (5′-UUUGUACUACACAAAAGUACUG-3′) were synthesized and obtained from Hippobio Co., Ltd. miR-193a-3p mimic, miR-146b-5p mimic and their mimic control (all 75 pM) were transfected into 2.5×105 293 cells/well using Lipofectamine® 3000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Transfection efficiency was measured by reverse transcription-quantitative PCR (RT-qPCR) after 48 h.

RNA extraction and RT-qPCR

Cellular RNA was extracted from 293 cells with TRIzol© reagent (Tiangen Biotech Co., Ltd.). RevertAid Reverse Transcriptase (Invitrogen; Thermo Fisher Scientific, Inc.) was used to reverse transcribe miRNA according to the manufacturer's protocol. The expression of miR-193a-3p and miR-146b-5p was performed by qPCR using PerfectStart Green qPCR SuperMix (Transgen Biotech Co., Ltd.) on an ABI Q1 RT-qPCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). U6 was used as an internal reference. The relevant primer sequences were as follows: miR-193a-3p forward, 5′-GCCGAGAACTGGCCTACAAA-3′ and reverse, 5′-CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGACTGGGAC-3′; miR-146b-5p forward, 5′-GCCGAGTGAGAACTGAATTCC-3′ and reverse, 5′-CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGCAGCCTAT-3′; U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′. miR-193a-3p and miR-146b-5p expression levels were calculated by the 2−∆∆Cq method (68).

Validation of the association between miR-193a-3p and the hub gene

The protein with the most connections was selected for further investigation. 293 cells exhibit a high transfection efficiency and were thus selected for the luciferase activity assay. The wild-type (Wt) CCND1-3′ untranslated region (UTR) and seed region mutant (Mut) of the miR-193a-3p binding site were subcloned into the pGL3 system (Promega Corporation). Its corresponding positive control [Wt tumor necrosis factor receptor-associated factor 6 (TRAF6)-3′ UTR + miR-146b-5p] was also subcloned into the pGL3 system (69,70). Subsequently, 1×105 293 cells/well were co-transfected with 100 ng Wt CCND1-3′ UTR or Wt TRAF6-3′ UTR vector and 400 ng miRNA mimic or negative control (NC) plasmid using X-tremeGENE™ HP DNA transfection reagent [Roche Diagnostics (Shanghai) Co., Ltd.]. Then, 2 days following incubation, firefly luciferase activity was assessed using a Dual-Luciferase Reporter assay system (Promega Corporation) in accordance with the manufacturer's protocols. Firefly luciferase activity was normalized to Renilla luciferase activity. Student's t-test was used to compare mimics and mimic control for each luciferase plasmid. Pearson's correlation analysis was also conducted to analyze the relationship between miR-193a-3p and CCND1, and P<0.05 was considered to indicate a statistically significant difference. Hub gene expression between the TC samples and the normal samples was explored via immunohistochemistry using The Human Protein Atlas (71).

Results

Downregulated miR-193a-3p expression in TC and its clinical significance from TCGA

The expression of miR-193a-3p in the TC group was 3.00±0.75 and that in the non-cancerous group was 3.88±0.73. miR-193a-3p exhibited significantly reduced expression in 510 TC samples compared with in 59 non-cancer controls (P<0.001; Fig. 1A). The ROC curve of miR-193a-3p was computed with an area under the curve (AUC) of 0.802 (P<0.001; Fig. 2A), which reflected the moderate value of miR-193a-3p to differentiate TC from the non-cancerous groups. The cut-off value was 3.444 (sensitivity 71.2% and specificity 79.7%). miR-193a-3p expression and clinical parameters are displayed in Table I. The miR-193a-3p levels in females were notably greater than those in males (P=0.004). miR-193a-3p was markedly decreased in individuals ≤60 years of age compared with those >60 years (P=0.020). ln addition, the number of distant metastases showed a negative association with miR-193a-3p expression levels (P=0.048).

Table I.

Association between miR-193a-3p expression and clinicopathological variables in TC from The Cancer Genome Atlas.

Table I.

Association between miR-193a-3p expression and clinicopathological variables in TC from The Cancer Genome Atlas.

miR-193a-3p expression

Clinicopathological featureCasesMean ± SDt/F valueP-value
Tissue
  Non-cancerous tissues593.88±0.738.468<0.001
  TC5103.00±0.75
Sex
  Male1342.89±0.73−2.022   0.044
  Female3683.04±0.76
Pathologic tumor grade
  Stage I–II3342.99±0.78−0.459   0.647
  Stage III–IV1663.02±0.72
Age
  <603833.04±0.752.336   0.020
  ≥601192.86±0.77
TNM stage
  T1-T23103.01±0.770.433   0.665
  T3-T41902.98±0.74
Pathological lymph node
  No2292.96±0.75−1.074   0.283
  Yes2233.04±0.73
Metastasis
  No2783.03±0.761.990   0.048
  Yes92.52±0.95
Ethnicity
  Caucasian3303.00±0.772.236   0.108
  African-American272.69±0.73
  Asian523.01±0.68

[i] miR, microRNA; TC, thyroid cancer.

miR-193a-3p expression in TC verified via GEO, ArrayExpress and literature analyses

Subsequently, in other online databases, data from 4 chips (GSE40807, GSE62054, GSE73182 and E-MTAB-736) were acquired from the GEO and ArrayExpress databases, which provided the expression information on miR-193a-3p in 88 TC and 75 non-cancerous tissues (Table II). The literature was also searched in 13 online databases, but the data from one study could not be extracted. All 4 datasets obtained from the GEO and ArrayExpress databases revealed that miR-193a-3p expression levels in TC were greater compared with non-TC controls, although two of these four datasets had P-values >0.05 (Fig. 1B-E). ROC analysis was also used to estimate the capacity of miR-193a-3p in distinguishing TC from non-cancer tissues (Fig. 2B-E); three of the four chips demonstrated that miR-193a-3p had a moderate diagnostic value for TC. miR-193a-3p expression data from TCGA, GEO and ArrayExpress, which provided 598 TC and 134 non-cancerous samples, were combined for meta-analysis. The SMD of miR-193a-3p was −1.00 [P<0.001; 95% CI, (−1.21, −0.79); Fig. 3A] via the fixed-effects model, and the heterogeneity test was P=0.46 (I2=0%). The AUC of the sROC was 0.80 (95% CI, 0.76–0.83), indicating a moderate ability to differentiate TC from non-cancer samples. The combined sensitivity and specificity were 0.69 (95% CI, 0.59–0.78) and 0.82 (95% CI, 0.67–0.91), respectively (Fig. 3B). Publication bias was performed and showed that there was no conspicuous publication bias among the datasets (Begg's test and Egger's test were 0.81 and 0.16, respectively; Fig. 3C and D).

Table II.

General characteristics of included microarray and RNA sequencing datasets.

Table II.

General characteristics of included microarray and RNA sequencing datasets.

Authors, yearCountryDatasetCancer groupNon-cancer groupsRefs.
TCGA, 2018USATCGA51059
Lassalle et al, 2016FranceGSE408074040(42)
Stokowy et al, 2014NorwayGSE62054175Unpublished data
Minna et al, 2016ItalyGSE73182195(43)
Rossing et al, 2012DenmarkE-MTAB-7361222(44)

[i] TCGA, The Cancer Genome Atlas.

GO annotation, KEGG pathway, DO and PPI network analysis

The prediction of the candidate target genes of miR-193a-3p was conducted using miRWalk 3.0, in which 3,984 potential mRNA candidate target genes were discovered. Next, 1,490 and 132 genes with elevated expression were determined from the TCGA and GEO databases, respectively. A total of 28 overlapping genes were eventually identified (Fig. 4A). The properties of these target genes were analyzed using the clusterprofiler R package. The results indicated that ‘response to nutrient levels’ was the most significantly enriched biological process (BP); with respect to cellular components (CC), the genes were mainly enriched in the ‘lamellar body’. In terms of molecular function (MF), ‘sulfur compound binding’ was the most enriched (Fig. 5A-C; Table III). However, no KEGG pathways were significantly enriched with these genes. DO analysis was also performed to discover related diseases caused by these genes. The results identified that ‘cell type benign neoplasm’ was the most enriched (Fig. 5D; Table III). Then, the target genes were put into the STRING online website to construct a protein-protein interaction network (Fig. 4B). In the present study, CCND1 attracted attention as a hub gene, as it had the most connections among the proteins; therefore, the targeting association between CCND1 and miR-193a-3p was selected for further investigation.

Table III.

Enrichment GO and disease ontology terms of the target genes of microRNA-193a-3p.

Table III.

Enrichment GO and disease ontology terms of the target genes of microRNA-193a-3p.

A, Biological processes

IDTermCountP-value
GO:0031667Response to nutrient levels  6 5.58×10−5
GO:0071900Regulation of protein serine/threonine kinase activity  6 6.50×10−5
GO:0009991Response to extracellular stimulus  6 8.06×10−5

B, Cellular component

IDTermCountP-value

GO:0042599Lamellar body  2 2.91×10−4
GO:0005771Multivesicular body  2 1.71×10−3
GO:0045178Basal part of cell  2 2.06×10−3

C, Molecular function

IDTermCountP-value

GO:1901681Sulfur compound binding  4 3.34×10−4
GO:0008201Heparin binding  3 1.46×10−3
GO:0098631Cell adhesion mediator activity  2 1.87×10−3

D, DO terms

IDTermCountP-value

DOID:0060084Cell type benign neoplasm10 2.79×10−7
DOID:1575Rheumatic disease  6 1.12×10−5
DOID:418Systemic scleroderma  6 1.12×10−5

[i] GO, Gene Ontology; DO, Disease Ontology.

Overexpression of CCND1 in TC and its clinical significance

The expression value and clinical significance of CCND1 in TC was verified using data from two different sources (TCGA and GEO databases; Table IV). The expression of CCND1 in the TC group from TCGA database was 13.36±0.74 and that in the non-cancerous group was 11.66±0.49. CCND1 was significantly overexpressed in 513 TC samples compared with 59 non-cancerous samples (P<0.001; Fig. 6A). The AUC for upregulated CCND1 expression in TC was 0.965 (P<0.001; Fig. 7A), which indicated the high capacity of the expression value of CCND1 to differentiate TC from normal samples. The cut-off value was 12.628 (sensitivity 86.9% and specificity 100%). Differential CCND1 expression was not detected for the clinical pathological parameters analyzed. The associations between the expression of CCND1 and clinical pathological parameters are summarized in Table V. The expression value of CCND1 was also calculated from the chip arrays of the GEO database, which included 384 TC and 316 non-cancer controls (GSE3678, GSE6004, GSE6339, GSE9115, GSE27115, GSE29265, GSE33630, GSE35570, GSE50901, GSE53072, GSE53157, GSE58689 and GSE65144). Except for GSE53072, GSE53157 and GSE65144, the remaining chip arrays revealed significantly upregulated expression of CCND1 in the TC tissues compared with non-cancerous tissues (P<0.05; Fig. 6B-N). The GSE53072, GSE53157 and GSE65144 chip arrays displayed non-significant trends towards upregulated expression of CCND1 in the TC tissues. AROC curve analysis of CCND1 was also performed and the results are shown in Fig. 7B-N. Then, CCND1 expression data from TCGA and GEO databases, which provided 897 TC and 193 non-cancerous tissues, were subjected to meta-analysis. The pooled SMD of CCND1 was 1.83 (P<0.001; 95% CI, 1.41–2.26; Fig. 8A) by the random effects model, and the test for heterogeneity was P<0.01 (I2=79.9%). The sROC curve of the TCGA and GEO expression data is shown in Fig. 8B. The publication bias was evaluated by a funnel chart: Begg's test and Egger's test were 0.44 and 0.75, respectively, which indicated that there was no conspicuous publication bias among the datasets (Fig. 8C and D). Furthermore, the AUC of the sROC was 0.91 (95% CI, 0.89–0.94) and the combined sensitivity and specificity were 0.85 (95% CI, 0.81–0.89) and 0.98 (95% CI, 0.90–1.00), respectively. The results revealed that CCND1 may be a good biomarker to discriminate TC tissues from non-cancerous tissues.

Table IV.

General characteristics of included microarray and RNA sequencing datasets.

Table IV.

General characteristics of included microarray and RNA sequencing datasets.

Authors, yearCountryDatasetCancer groupNon-cancer groupsRefs.
TCGA, 2018USATCGA51359
Reyes et al, 2005USAGSE367877Unpublished data
Vasko et al, 2007USAGSE6004144(57)
Fontaine et al, 2008FranceGSE633920122(58)
Salvatore et al, 2007USAGSE9115154(59)
Giordano et al, 2006USAGSE271557821(60)
Tomas et al, 2011BelgiumGSE292652920Unpublished data
Dom et al, 2012BelgiumGSE336306045(61)
Handkiewicz-Junak et al, 2016PolandGSE355703251(62)
Barros-Filho et al, 2015BrazilGSE50901614(63)
Pita et al, 2014PortugalGSE5307254(64)
Pita et al, 2009PortugalGSE53157243(65)
Rusinek et al, 2015PolandGSE586892718(66)
von Roemeling et al, 2015USAGSE651441213(67)

[i] TCGA, The Cancer Genome Atlas.

Table V.

Clinical parameters and CCND1 expression in TC from The Cancer Genome Atlas.

Table V.

Clinical parameters and CCND1 expression in TC from The Cancer Genome Atlas.

CCND1 expression

Clinicopathological featureCasesMean ± SDt/F valueP-value
Tissue
  Non-cancerous tissues5911.66±0.49−23.800<0.001
  TC51313.36±0.74
Sex
  Male13613.33±0.80   −0.574   0.566
  Female36913.37±0.72
Pathologic tumor grade
  Stage I–II33613.33±0.77   −1.269   0.205
  Stage III–IV16713.42±0.68
Age
  <6038513.38±0.73   1.114   0.266
  ≥6012013.30±0.78
TNM stage
  T1-T230913.33±0.74   −1.148   0.251
  T3-T419413.41±0.74
Pathological lymph node
  No23013.32±0.84   −1.849   0.065
  Yes22513.44±0.55
Metastasis
  No28213.40±0.63   −0.662   0.508
  Yes913.55±0.71
Ethnicity
  Caucasian33413.38±0.69   1.278   0.280
  African-American2713.58±0.43
  Asian5113.45±0.73

[i] CCND1, cyclin D1; TC, thyroid cancer.

Validation of the targeting regulatory relation between miR-193a-3p and CCND1

The putative miR-193a-3p binding site in CCND1 3′ UTR was identified (Fig. 9A). The targeting regulatory association between miR-193a-3p and CCND1 was investigated in 293 cells following transfection with miR-193a-3p or miR-146b-5p mimics. The findings indicated that miR-193a-3p reduced the luciferase activity of the Wt CCND1 3′ UTR, demonstrating a direct combination between CCND1 and miR-193a-3p (P<0.05; Fig. 9B-D). Furthermore, the luciferase activity of CCND1 3′ UTR (Mut) was not reduced in the mimic-transfected cells. miR-146-5p reduced the luciferase activity of the Wt TRAF6-3′ UTR, indicating that the experiment and its results were reliable. Pearson correlation analysis was also conducted to evaluate the association between miR-193a-3p and CCND1; however, no linear correlation was observed between CCND1 and miR-193a-3p (r=−0.163; Fig. 9E). The Human Protein Atlas database was utilized to explore the protein levels of CCND1. Consistent with the gene expression data, CCND1 protein expression was higher in TC than in non-cancerous tissues, as shown in Fig. 10.

Discussion

TC is a frequent endocrine malignancy of the endocrine system (72). The occurrence rate of TC has been increasing worldwide (73). The main therapy method of TC is thyroidectomy and radioiodine treatment (74). To the best of the authors' knowledge, there is currently no effective drug for the treatment for TC. There is increasing evidence that abnormal miRNA expression levels give rise to TC tumorigenesis and progression. Prior studies have demonstrated that various miRNAs may aid in the diagnosis of TC and help formulate more individualized therapeutic management strategies (7578). miR-193a-3p is involved in certain biological processes, including apoptosis, proliferation, invasion, metastasis and migration (79). A number of studies have demonstrated that miR-193a-3p is expressed at low levels in different human cancers, including colon cancer (80), lung cancer (81,82), breast cancer (28) and malignant pleural mesothelioma (83). Nevertheless, the expression levels, as well as its mechanisms, in TC remain to be elucidated. Hence, a comprehensive meta-analysis was conducted to evaluate clinical valuation of miR-193a-3p in TC. GO, KEGG pathway and PPI analyses were performed to search for the latent biological mechanisms of miR-193a-3p in TC. The results indicated that CCND1 served as the most important hub gene. Subsequently, the relationship between miR-193a-3p and CCND1 was validated through a dual luciferase assay.

To the best of the authors' knowledge, the present study is the first to survey the expression levels and clinical value of miR-193a-3p in TC by extracting expression data from TCGA, GEO and ArrayExpress. The present study primarily scrutinized the miR-193a-3p expression between 510 TC and 59 non-cancerous thyroid samples using TCGA database. The outcomes revealed that miR-193a-3p had low expression in TC. Subsequently, the GEO database and ArrayExpress were used to validate miR-193a-3p expression in TC. A total of 2 of the 4 chip arrays also revealed that a low miR-193a-3p expression is significant in TC. Although the remaining 2 chip arrays did not display a significant downregulation in TC tissues, they indicated the downregulated expression tendency of miR-193a-3p in TC. According to the results of the present study, miR-193a-3p may provide a biomarker to test the occurrence and development of TC. However, in the study by Santarpia et al (31), the result was the opposite: miR-193a-3p was overexpressed in TC compared with non-cancerous groups. The observation of overexpressed miR-193a-3p may be caused by the small sample size of the study. That significantly downregulated miR-193a-3p in TC was observed in the present study was credible due to the quantity of sample data that was obtained from several public databases. Analysis of the sROC in miR-193a-3p from TCGA, GEO and ArrayExpress databases demonstrated that the expression levels may have a moderate ability to differentiate between TC patients and non-cancerous controls with an AUC of 0.80. At the same time, its sensitivity and specificity were 0.69 and 0.82, respectively. Additionally, downregulated miR-193a-3p was association with age, metastasis and sex in TCGA. Although the present study identified that downregulated miR-193a-3p was associated with age, metastasis and sex; how these factors regulate TC cells requires further investigation by in vivo and/or in vitro analysis.

At present, several studies have noted that abnormal miR-193a-3p expression has a pivotal effect on cell biological processes and leads to numerous pathological conditions (79,80,84). For example, Pekow et al revealed that miR-193a-3p is a crucial neoplasm inhibitor in colon carcinoma and facilitates the progression of cancer (80). Lin et al (84). demonstrated that low miR-193a-3p expression is related to a poor disease outcome in colorectal carcinoma. In addition, miR-193a-3p suppresses cell multiplication in prostatic carcinoma by targeting CCND1 (85). However, little research has been performed into the molecular mechanisms of miR-193a-3p in TC. To further study the underlying mechanisms regarding the regulation of miR-193a-3p in TC, the present study conducted a bioinformatics analysis of the candidate target genes of miR-193a-3p. A total of 28 candidate target genes of miR-193a-3p were gathered and GO analysis was performed, which indicated that the candidate target genes markedly assembled in ‘response to nutrient levels’ for BP, ‘lamellar body’ for CC and ‘sulfur compound binding’ for MF. GO analysis revealed that miR-193a-3p target genes were mainly concentrated in ‘cell adhesion’, which exerts an oncogenic role in the progression of TC. In addition, PPI analysis was performed, and it was found that CCND1 was situated at the core of the PPI network. CCND1 had the most connections among proteins, which may manifest in the way the gene contributes to TC.

A previous study by Li et al (35) reported that CCND1 plays a key role in facilitating migration and tumor metastasis. Liang and Sun (86) reported that an upregulation of CCND1 in thyroid cancer tissues is markedly associated with the clinical stage of TC, indicating that it could be considered as a biomarker in clinical settings. A study by Jeon et al (87) showed that CCND1 is clearly overexpressed in TC, and may be involved in the occurrence and development of the disease. Lamba Saini et al (88) demonstrated that CCND1 and its variants are significantly overexpressed in TC, and that it should serve as a diagnostic marker for TC. Investigating the mechanism of CCND1 expression levels is crucial due to the broader roles of CCND1 in TC tissues. From the present study, the results calculated from the TCGA database demonstrated that CCND1 exhibited a notably low expression in TC compared with non-cancerous tissues. However, differential expression of CCND1 was not obtained for the clinical parameters analyzed. The present study also included 13 microarrays from the GEO database. In addition to GSE53072, GSE53157, GSE6339 and GSE65144, other chip arrays all displayed a significant upregulation of CCND1 expression in TC compared to non-cancerous tissues. Although the remaining 4 chip arrays did not show statistical significance, they all showed an increasing trend in CCND1 expression levels in TC. From the sROC analysis of miR-193a-3p, the AUC was 0.91. The results showed that CCND1 may have a high value for differentiating TC from non-cancerous tissue and may serve as a prognostic biomarker in TC.

The study by Liu et al (85) identified that miR-193a-3p acts as a key component inhibiting prostate cancer, which could induce G1-phase arrest by targeting CCND1. Chou et al (25) demonstrated that miR-193a-3p suppresses the aggression and progression of gastric cancer by targeting CCND1 expression. Tsai et al (89) also demonstrated that miR-193a-3p may serve as an underlying neoplasm inhibitor in breast carcinoma, which could inhibit cell growth by suppressing CCND1. To this end, the present study performed a dual luciferase assay, and a regulatory association between miR-193a-3p and CCND1 was validated. In summary, the findings of the present study revealed that miR-193a-3p may regulate CCND1 expression to affect the tumor cell cycle, cell proliferation, differentiation and metabolism. Nevertheless, further research is required to test this hypothesis.

However, in the present study, some limitations should be acknowledged. First, it lacks further in vitro and in vivo testing, such as reverse transcription-quantitative PCR. These methods are pivotal to enhancing the understanding of how miR-193a-3p works in relation to CCND1 in TC. Second, the present study only focused on the analysis of differential miRNAs in tissues. Other samples, such as blood, should also be assessed.

In summary, the present study gathered a great amount of data from TCGA, GEO, and ArrayExpress databases, and validated the clinical value of miR-193a-3p and CCND1. The results showed that miR-193a-3p expression was evidently downregulated in TC, while CCND1 expression was markedly upregulated. These results may help to diagnose TC and predict the prognosis of TC when thyroid tissues are obtained from a fine needle biopsy. Bioinformatics analysis was next performed to clarify the molecular mechanisms of miR-193a-3p. In light of the bioinformatics analysis, CCND1 was regarded as the most important hub gene in TC. Subsequently, a dual luciferase assay was performed to corroborate the targeting regulatory association between miR-193a-3p and CCND1. The results demonstrated that miR-193a-3p markedly contributes to TC via particular pathways, and both miR-193a-3p and CCND1 may serve as potential biological markers of TC.

Acknowledgements

Not applicable.

Funding

This study was supported by Fund of National Natural Science Foundation of China (grant nos. NSFC81060202 and NSFC81260222), the Fund of Guangxi Key R&D Project Plan (grant no. AB17195020) and the Future Academic Star of Guangxi Medical University (grant nos. WLXSZX19050 and WLXSZX19055).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YYP, HY, XJL and RW contributed to the design of the study; XJL and RW wrote the manuscript; DYW, PL, DHP, LJZ and YH collected the data, performed the statistical analysis and interpreted the data; and LS, YYQ, YHL, JNL, JLY, QQL, JW and JM contributed to the data collection and statistical analysis. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Konturek A, Barczyński M, Stopa M and Nowak W: Trends in Prevalence of Thyroid Cancer Over Three Decades: A Retrospective Cohort Study of 17,526 Surgical Patients. World J Surg. 40:538–544. 2016. View Article : Google Scholar : PubMed/NCBI

2 

van der Zwan JM, Mallone S, van Dijk B, Bielska-Lasota M, Otter R, Foschi R, Baudin E and Links TP; RARECARE WG, : Carcinoma of endocrine organs: Results of the RARECARE project. Eur J Cancer. 48:1923–1931. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Miller KD, Goding Sauer A, Ortiz AP, Fedewa SA, Pinheiro PS, Tortolero-Luna G, Martinez-Tyson D, Jemal A and Siegel RL: Cancer Statistics for Hispanics/Latinos, 2018. CA Cancer J Clin. 68:425–445. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Lin P, Guo YN, Shi L, Li XJ, Yang H, He Y, Li Q, Dang YW, Wei KL and Chen G: Development of a prognostic index based on an immunogenomic landscape analysis of papillary thyroid cancer. Aging (Albany NY). 11:480–500. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Zarkesh M, Zadeh-Vakili A, Akbarzadeh M, Fanaei SA, Hedayati M and Azizi F: The role of matrix metalloproteinase-9 as a prognostic biomarker in papillary thyroid cancer. BMC Cancer. 18:11992018. View Article : Google Scholar : PubMed/NCBI

6 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Carling T and Udelsman R: Thyroid cancer. Annu Rev Med. 65:125–137. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Lin P, He Y, Wen DY, Li XJ, Zeng JJ, Mo WJ, Li Q, Peng JB, Wu YQ, Pan DH, et al: Comprehensive analysis of the clinical significance and prospective molecular mechanisms of differentially expressed autophagy-related genes in thyroid cancer. Int J Oncol. 53:603–619. 2018.PubMed/NCBI

9 

Liu C, Su C, Chen Y and Li G: miR-144-3p promotes the tumor growth and metastasis of papillary thyroid carcinoma by targeting paired box gene 8. Cancer Cell Int. 18:542018. View Article : Google Scholar : PubMed/NCBI

10 

Acquaviva G, Visani M, Repaci A, Rhoden KJ, de Biase D, Pession A and Giovanni T: Molecular pathology of thyroid tumours of follicular cells: A review of genetic alterations and their clinicopathological relevance. Histopathology. 72:6–31. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Riesco-Eizaguirre G and Santisteban P: Molecular biology of thyroid cancer initiation. Clin Transl Oncol. 9:686–693. 2007. View Article : Google Scholar : PubMed/NCBI

12 

DeLellis RA: Pathology and genetics of thyroid carcinoma. J Surg Oncol. 94:662–669. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Liu T, You X, Sui J, Shen B, Zhang Y, Zhang XM, Yang S, Yao YZ, Yang F, Yin LH, et al: Prognostic value of a two-microRNA signature for papillary thyroid cancer and a bioinformatic analysis of their possible functions. J Cell Biochem. Nov 2–2018.(Epub ahead of print). doi: 10.1002/jcb.27993 2018.

14 

Wang X, Huang S, Li X, Jiang D, Yu H, Wu Q, Gao C and Wu Z: A potential biomarker hsa-miR-200a-5p distinguishing between benign thyroid tumors with papillary hyperplasia and papillary thyroid carcinoma. PLoS One. 13:e02002902018. View Article : Google Scholar : PubMed/NCBI

15 

Vuong HG, Altibi AM, Abdelhamid AH, Ngoc PU, Quan VD, Tantawi MY, Elfil M, Vu TL, Elgebaly A, Oishi N, et al: The changing characteristics and molecular profiles of papillary thyroid carcinoma over time: A systematic review. Oncotarget. 8:10637–10649. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Tricoli JV and Jacobson JW: MicroRNA: Potential for Cancer Detection, Diagnosis, and Prognosis. Cancer Res. 67:4553–4555. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Boufraqech M, Klubo-Gwiezdzinska J and Kebebew E: MicroRNAs in the thyroid. Best Pract Res Clin Endocrinol Metab. 30:603–619. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Macha MA, Seshacharyulu P, Krishn SR, Pai P, Rachagani S, Jain M and Batra SK: MicroRNAs (miRNAs) as biomarker(s) for prognosis and diagnosis of gastrointestinal (GI) cancers. Curr Pharm Des. 20:5287–5297. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Parvex P: Are microRNA potential biomarkers in children with idiopathic nephrotic syndrome? EBioMedicine. 39:27–28. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Farazi TA, Spitzer JI, Morozov P and Tuschl T: miRNAs in human cancer. J Pathol. 223:102–115. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Li Z, Yu X, Shen J, Law PT, Chan MT and Wu WK: MicroRNA expression and its implications for diagnosis and therapy of gallbladder cancer. Oncotarget. 6:13914–13921. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Li L, Peng M, Xue W, Fan Z, Wang T, Lian J, Zhai Y, Lian W, Qin D and Zhao J: Integrated analysis of dysregulated long non-coding RNAs/microRNAs/mRNAs in metastasis of lung adenocarcinoma. J Transl Med. 16:3722018. View Article : Google Scholar : PubMed/NCBI

23 

Ma Y and Sun Y: miR-29a-3p inhibits growth, proliferation, and invasion of papillary thyroid carcinoma by suppressing NF-κB signaling via direct targeting of OTUB2. Cancer Manag Res. 11:13–23. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Liu Y, Ren F, Luo Y, Rong M, Chen G and Dang Y: Down-Regulation of miR-193a-3p Dictates Deterioration of HCC: A Clinical Real-Time qRT-PCR Study. Med Sci Monit. 21:2352–2360. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Chou NH, Lo YH, Wang KC, Kang CH, Tsai CY and Tsai KW: miR-193a-5p and −3p Play a Distinct Role in Gastric Cancer: miR-193a-3p Suppresses Gastric Cancer Cell Growth by Targeting ETS1 and CCND1. Anticancer Res. 38:3309–3318. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Huang Y, Luo H, Li F, Yang Y, Ou G, Ye X and Li N: LINC00152 down-regulated miR-193a-3p to enhance MCL1 expression and promote gastric cancer cells proliferation. Biosci Rep. 38:BSR201716072018. View Article : Google Scholar : PubMed/NCBI

27 

Deng W, Yan M, Yu T, Ge H, Lin H, Li J, Liu Y, Geng Q, Zhu M, Liu L, et al: Quantitative proteomic analysis of the metastasis-inhibitory mechanism of miR-193a-3p in non-small cell lung cancer. Cell Physiol Biochem. 35:1677–1688. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Yu M, Liu Z, Liu Y, Zhou X, Sun F, Liu Y, Li L, Hua S, Zhao Y, Gao H, et al: PTP1B markedly promotes breast cancer progression and is regulated by miR-193a-3p. FEBS J. 286:1136–1153. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Takahashi H, Takahashi M, Ohnuma S, Unno M, Yoshino Y, Ouchi K, Takahashi S, Yamada Y, Shimodaira H and Ishioka C: microRNA-193a-3p is specifically down-regulated and acts as a tumor suppressor in BRAF-mutated colorectal cancer. BMC Cancer. 17:7232017. View Article : Google Scholar : PubMed/NCBI

30 

Mamoori A, Wahab R, Islam F, Lee K, Vider J, Lu CT, Gopalan V and Lam AK: Clinical and biological significance of miR-193a-3p targeted KRAS in colorectal cancer pathogenesis. Hum Pathol. 71:145–156. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Santarpia L, Calin GA, Adam L, Ye L, Fusco A, Giunti S, Thaller C, Paladini L, Zhang X, Jimenez C, et al: A miRNA signature associated with human metastatic medullary thyroid carcinoma. Endocr Relat Cancer. 20:809–823. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Baldin V, Lukas J, Marcote MJ, Pagano M and Draetta G: Cyclin D1 is a nuclear protein required for cell cycle progression in G1. Genes Dev. 7:812–821. 1993. View Article : Google Scholar : PubMed/NCBI

33 

Zhao M, Xu P, Liu Z, Zhen Y, Chen Y, Liu Y, Fu Q, Deng X, Liang Z, Li Y, et al: Dual roles of miR-374a by modulated c-Jun respectively targets CCND1-inducing PI3K/AKT signal and PTEN-suppressing Wnt/β-catenin signaling in non-small-cell lung cancer. Cell Death Dis. 9:782018. View Article : Google Scholar : PubMed/NCBI

34 

Li Y, Li D, Yang W, Fu H, Liu Y and Li Y: Overexpression of the transcription factor FOXP3 in lung adenocarcinoma sustains malignant character by promoting G1/S transition gene CCND1. Tumour Biol. 37:7395–7404. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Li Z, Wang C, Prendergast GC and Pestell RG: Cyclin D1 functions in cell migration. Cell Cycle. 5:2440–2442. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Huang H, Han Y, Yang X, Li M, Zhu R, Hu J, Zhang X, Wei R, Li K and Gao R: HNRNPK inhibits gastric cancer cell proliferation through p53/p21/CCND1 pathway. Oncotarget. 8:103364–103374. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Xue J, Qin Z, Li X, Zhang J, Zheng Y, Xu W, Cao Q and Wang Z: Genetic polymorphisms in cyclin D1 are associated with risk of renal cell cancer in the Chinese population. Oncotarget. 8:80889–80899. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Cheng S, Serra S, Mercado M, Ezzat S and Asa SL: A high-throughput proteomic approach provides distinct signatures for thyroid cancer behavior. Clin Cancer Res. 17:2385–2394. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Deng M, Brägelmann J, Schultze JL and Perner S: Web-TCGA: An online platform for integrated analysis of molecular cancer data sets. BMC Bioinformatics. 17:722016. View Article : Google Scholar : PubMed/NCBI

40 

Chandran UR, Medvedeva OP, Barmada MM, Blood PD, Chakka A, Luthra S, Ferreira A, Wong KF, Lee AV, Zhang Z, et al: TCGA Expedition: A Data Acquisition and Management System for TCGA Data. PLoS One. 11:e01653952016. View Article : Google Scholar : PubMed/NCBI

41 

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al: NCBI GEO: Archive for functional genomics data sets - update. Nucleic Acids Res 41D. D991–D995. 2013.

42 

Lassalle S, Zangari J, Popa A, Ilie M, Hofman V, Long E, Patey M, Tissier F, Belléannée G, Trouette H, et al: MicroRNA-375/SEC23A as biomarkers of the in vitro efficacy of vandetanib. Oncotarget. 7:30461–30478. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Minna E, Romeo P, Dugo M, De Cecco L, Todoerti K, Pilotti S, Perrone F, Seregni E, Agnelli L, Neri A, et al: miR-451a is underexpressed and targets AKT/mTOR pathway in papillary thyroid carcinoma. Oncotarget. 7:12731–12747. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Rossing M, Borup R, Henao R, Winther O, Vikesaa J, Niazi O, Godballe C, Krogdahl A, Glud M, Hjort-Sørensen C, et al: Down-regulation of microRNAs controlling tumourigenic factors in follicular thyroid carcinoma. J Mol Endocrinol. 48:11–23. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Ioannidis JP, Patsopoulos NA and Evangelou E: Uncertainty in heterogeneity estimates in meta-analyses. BMJ. 335:914–916. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Gan BL, He RQ, Zhang Y, Wei DM, Hu XH and Chen G: Downregulation of HOXA3 in lung adenocarcinoma and its relevant molecular mechanism analysed by RT-qPCR, TCGA and in silico analysis. Int J Oncol. 53:1557–1579. 2018.PubMed/NCBI

47 

Deng Y, He R, Zhang R, Gan B, Zhang Y, Chen G and Hu X: The expression of HOXA13 in lung adenocarcinoma and its clinical significance: A study based on The Cancer Genome Atlas, Oncomine and reverse transcription-quantitative polymerase chain reaction. Oncol Lett. 15:8556–8572. 2018.PubMed/NCBI

48 

Liang YY, Huang JC, Tang RX, Chen WJ, Chen P, Cen WL, Shi K, Gao L, Gao X, Liu AG, et al: Clinical value of miR-198-5p in lung squamous cell carcinoma assessed using microarray and RT-qPCR. World J Surg Oncol. 16:222018. View Article : Google Scholar : PubMed/NCBI

49 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

50 

Bardou P, Mariette J, Escudié F, Djemiel C and Klopp C: jvenn: An interactive Venn diagram viewer. BMC Bioinformatics. 15:2932014. View Article : Google Scholar : PubMed/NCBI

51 

The Gene Ontology Consortium, . The Gene Ontology Resource: 20 years and still GOing strong. Nucleic Acids Res. 47(D1): D330–D338. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Kanehisa M, Sato Y, Furumichi M, Morishima K and Tanabe M: New approach for understanding genome variations in KEGG. Nucleic Acids Res 47D. D590–D595. 2019. View Article : Google Scholar

53 

Schriml LM, Mitraka E, Munro J, Tauber B, Schor M, Nickle L, Felix V, Jeng L, Bearer C, Lichenstein R, et al: Human Disease Ontology 2018 update: Classification, content and workflow expansion. Nucleic Acids Res 47D. D955–D962. 2019. View Article : Google Scholar

54 

Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Wickham H: ggplot2: Elegant Graphics for Data Analysis. Springer; New York, NY: 2016

56 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 47D. D607–D613. 2019. View Article : Google Scholar

57 

Vasko V, Espinosa AV, Scouten W, He H, Auer H, Liyanarachchi S, Larin A, Savchenko V, Francis GL, de la Chapelle A, et al: Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion. Proc Natl Acad Sci USA. 104:2803–2808. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Fontaine JF, Mirebeau-Prunier D, Franc B, Triau S, Rodien P, Houlgatte R, Malthièry Y and Savagner F: Microarray analysis refines classification of non-medullary thyroid tumours of uncertain malignancy. Oncogene. 27:2228–2236. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Salvatore G, Nappi TC, Salerno P, Jiang Y, Garbi C, Ugolini C, Miccoli P, Basolo F, Castellone MD, Cirafici AM, et al: A cell proliferation and chromosomal instability signature in anaplastic thyroid carcinoma. Cancer Res. 67:10148–10158. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Giordano TJ, Au AY, Kuick R, Thomas DG, Rhodes DR, Wilhelm KG Jr, Vinco M, Misek DE, Sanders D, Zhu Z, et al: Delineation, functional validation, and bioinformatic evaluation of gene expression in thyroid follicular carcinomas with the PAX8-PPARG translocation. Clin Cancer Res. 12:1983–1993. 2006. View Article : Google Scholar : PubMed/NCBI

61 

Dom G, Tarabichi M, Unger K, Thomas G, Oczko-Wojciechowska M, Bogdanova T, Jarzab B, Dumont JE, Detours V and Maenhaut C: A gene expression signature distinguishes normal tissues of sporadic and radiation-induced papillary thyroid carcinomas. Br J Cancer. 107:994–1000. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Handkiewicz-Junak D, Swierniak M, Rusinek D, Oczko-Wojciechowska M, Dom G, Maenhaut C, Unger K, Detours V, Bogdanova T, Thomas G, et al: Gene signature of the post-Chernobyl papillary thyroid cancer. Eur J Nucl Med Mol Imaging. 43:1267–1277. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Barros-Filho MC, Marchi FA, Pinto CA, Rogatto SR and Kowalski LP: High Diagnostic Accuracy Based on CLDN10, HMGA2, and LAMB3 Transcripts in Papillary Thyroid Carcinoma. J Clin Endocrinol Metab. 100:E890–E899. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Pita JM, Figueiredo IF, Moura MM, Leite V and Cavaco BM: Cell cycle deregulation and TP53 and RAS mutations are major events in poorly differentiated and undifferentiated thyroid carcinomas. J Clin Endocrinol Metab. 99:E497–E507. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Pita JM, Banito A, Cavaco BM and Leite V: Gene expression profiling associated with the progression to poorly differentiated thyroid carcinomas. Br J Cancer. 101:1782–1791. 2009. View Article : Google Scholar : PubMed/NCBI

66 

Rusinek D, Swierniak M, Chmielik E, Kowal M, Kowalska M, Cyplinska R, Czarniecka A, Piglowski W, Korfanty J, Chekan M, et al: BRAFV600E-Associated Gene Expression Profile: Early Changes in the Transcriptome, Based on a Transgenic Mouse Model of Papillary Thyroid Carcinoma. PLoS One. 10:e01436882015. View Article : Google Scholar : PubMed/NCBI

67 

von Roemeling CA, Marlow LA, Pinkerton AB, Crist A, Miller J, Tun HW, Smallridge RC and Copland JA: Aberrant lipid metabolism in anaplastic thyroid carcinoma reveals stearoyl CoA desaturase 1 as a novel therapeutic target. J Clin Endocrinol Metab. 100:E697–E709. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

69 

Liu J, Xu J, Li H, Sun C, Yu L, Li Y, Shi C, Zhou X, Bian X, Ping Y, et al: miR-146b-5p functions as a tumor suppressor by targeting TRAF6 and predicts the prognosis of human gliomas. Oncotarget. 6:29129–29142. 2015. View Article : Google Scholar : PubMed/NCBI

70 

Xiong DD, Li ZY, Liang L, He RQ, Ma FC, Luo DZ, Hu XH and Chen G: The LncRNA NEAT1 Accelerates Lung Adenocarcinoma Deterioration and Binds to Mir-193a-3p as a Competitive Endogenous RNA. Cell Physiol Biochem. 48:905–918. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, et al: Proteomics. Tissue-based map of the human proteome. Science. 347:12604192015. View Article : Google Scholar : PubMed/NCBI

72 

Kitahara CM and Sosa JA: The changing incidence of thyroid cancer. Nat Rev Endocrinol. 12:646–653. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Vigneri R, Malandrino P and Vigneri P: The changing epidemiology of thyroid cancer: Why is incidence increasing? Curr Opin Oncol. 27:1–7. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Raue F and Frank-Raue K: Thyroid Cancer: Risk-Stratified Management and Individualized Therapy. Clin Cancer Res. 22:5012–5021. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Qiu J, Zhang W, Zang C, Liu X, Liu F, Ge R, Sun Y and Xia Q: Identification of key genes and miRNAs markers of papillary thyroid cancer. Biol Res. 51:452018. View Article : Google Scholar : PubMed/NCBI

76 

Nixon AM, Provatopoulou X, Kalogera E, Zografos GN and Gounaris A: Circulating thyroid cancer biomarkers: Current limitations and future prospects. Clin Endocrinol (Oxf). 87:117–126. 2017. View Article : Google Scholar : PubMed/NCBI

77 

Zhang Y, Pan J, Xu D, Yang Z, Sun J, Sun L, Wu Y and Qiao H: Combination of serum microRNAs and ultrasound profile as predictive biomarkers of diagnosis and prognosis for papillary thyroid microcarcinoma. Oncol Rep. 40:3611–3624. 2018.PubMed/NCBI

78 

Nikiforov YE: Role Of Molecular Markers In Thyroid Nodule Management: Then And Now. Endocr Pract. 23:979–988. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Grossi I, Salvi A, Abeni E, Marchina E and De Petro G: Biological Function of MicroRNA193a-3p in Health and Disease. Int J Genomics. 2017:59131952017. View Article : Google Scholar : PubMed/NCBI

80 

Pekow J, Meckel K, Dougherty U, Huang Y, Chen X, Almoghrabi A, Mustafi R, Ayaloglu-Butun F, Deng Z, Haider HI, et al: miR-193a-3p is a Key Tumor Suppressor in Ulcerative Colitis-Associated Colon Cancer and Promotes Carcinogenesis through Upregulation of IL17RD. Clin Cancer Res. 23:5281–5291. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Fan Q, Hu X, Zhang H, Wang S, Zhang H, You C, Zhang CY, Liang H, Chen X and Ba Y: miR-193a-3p is an Important Tumour Suppressor in Lung Cancer and Directly Targets KRAS. Cell Physiol Biochem. 44:1311–1324. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Ren F, Ding H, Huang S, Wang H, Wu M, Luo D, Dang Y, Yang L and Chen G: Expression and clinicopathological significance of miR-193a-3p and its potential target astrocyte elevated gene-1 in non-small lung cancer tissues. Cancer Cell Int. 15:802015. View Article : Google Scholar : PubMed/NCBI

83 

Williams M, Kirschner MB, Cheng YY, Hanh J, Weiss J, Mugridge N, Wright CM, Linton A, Kao SC, Edelman JJ, et al: miR-193a-3p is a potential tumor suppressor in malignant pleural mesothelioma. Oncotarget. 6:23480–23495. 2015. View Article : Google Scholar : PubMed/NCBI

84 

Lin M, Duan B, Hu J, Yu H, Sheng H, Gao H and Huang J: Decreased expression of miR-193a-3p is associated with poor prognosis in colorectal cancer. Oncol Lett. 14:1061–1067. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Liu Y, Xu X, Xu X, Li S, Liang Z, Hu Z, Wu J, Zhu Y, Jin X, Wang X, et al: MicroRNA-193a-3p inhibits cell proliferation in prostate cancer by targeting cyclin D1. Oncol Lett. 14:5121–5128. 2017.PubMed/NCBI

86 

Liang W and Sun F: Identification of key genes of papillary thyroid cancer using integrated bioinformatics analysis. J Endocrinol Invest. 41:1237–1245. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Jeon S, Kim Y, Jeong YM, Bae JS and Jung CK: CCND1 Splice Variant as A Novel Diagnostic and Predictive Biomarker for Thyroid Cancer. Cancers (Basel). 10:4372018. View Article : Google Scholar

88 

Lamba Saini M, Weynand B, Rahier J, Mourad M, Hamoir M and Marbaix E: Cyclin D1 in well differentiated thyroid tumour of uncertain malignant potential. Diagn Pathol. 10:322015. View Article : Google Scholar : PubMed/NCBI

89 

Tsai KW, Leung CM, Lo YH, Chen TW, Chan WC, Yu SY, Tu YT, Lam HC, Li SC, Ger LP, et al: Arm Selection Preference of MicroRNA-193a Varies in Breast Cancer. Sci Rep. 6:281762016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 22 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li XJ, Wen R, Wen DY, Lin P, Pan DH, Zhang LJ, He Y, Shi L, Qin YY, Lai YH, Lai YH, et al: Downregulation of miR‑193a‑3p via targeting cyclin D1 in thyroid cancer. Mol Med Rep 22: 2199-2218, 2020
APA
Li, X., Wen, R., Wen, D., Lin, P., Pan, D., Zhang, L. ... Pang, Y. (2020). Downregulation of miR‑193a‑3p via targeting cyclin D1 in thyroid cancer. Molecular Medicine Reports, 22, 2199-2218. https://doi.org/10.3892/mmr.2020.11310
MLA
Li, X., Wen, R., Wen, D., Lin, P., Pan, D., Zhang, L., He, Y., Shi, L., Qin, Y., Lai, Y., Lai, J., Yang, J., Lai, Q., Wang, J., Ma, J., Yang, H., Pang, Y."Downregulation of miR‑193a‑3p via targeting cyclin D1 in thyroid cancer". Molecular Medicine Reports 22.3 (2020): 2199-2218.
Chicago
Li, X., Wen, R., Wen, D., Lin, P., Pan, D., Zhang, L., He, Y., Shi, L., Qin, Y., Lai, Y., Lai, J., Yang, J., Lai, Q., Wang, J., Ma, J., Yang, H., Pang, Y."Downregulation of miR‑193a‑3p via targeting cyclin D1 in thyroid cancer". Molecular Medicine Reports 22, no. 3 (2020): 2199-2218. https://doi.org/10.3892/mmr.2020.11310